Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Prog Pediatr Cardiol ; 67: 101549, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35813038

RESUMEN

Background: The COVID pandemic necessitated an altered approach to transthoracic echocardiography, especially in COVID cases. Whether this has effected echocardiography lab quality is unknown. Objectives: We sought to determine whether echocardiography lab quality measures during the COVID pandemic were different from those prior to the pandemic and whether quality and comprehensiveness of echocardiograms performed during the pandemic was different between COVID and non-COVID patients. Methods: The four quality measures (diagnostic errors, appropriateness of echocardiogram, American College of Cardiology Image Quality metric and Comprehensive Exam metric in structurally normal hearts) reported quarterly in our lab were compared between two quarters during COVID (2020) and pre-COVID (2019). Each component of these metrics was also assessed in randomly selected echocardiograms in COVID patients and compared to non-COVID echocardiograms. Results: For non-COVID echocardiograms, the image quality metric did not change between 2019 and 2020 and the comprehensive exam metric improved. Diagnostic error rate did not change, and appropriateness of echocardiogram indications improved. When COVID and non-COVID echocardiograms were compared, the image quality metric and comprehensiveness exam metric were lower for COVID cases (image quality mean 21.3/23 for non-COVID, 18.6/23 for COVID, p < 0.001 and comprehensive exam mean 29.5/30 for non-COVID, 27.7/39 for COVID, p < 0.001). In particular, systemic and pulmonary veins, pulmonary arteries and aortic arch were not adequately imaged in COVID patients. For studies in which a follow-up echocardiogram was available, no new pathology was found. Conclusions: At our center, though diagnostic error rate did not change during the pandemic and the proportion of echocardiograms ordered for appropriate indications increased, imaging quality in COVID patients was compromised, especially for systemic and pulmonary veins, pulmonary arteries and arch. Though no new pathology was noted on the small number of patients who had follow-up studies, we are paying careful attention to these structures to avoid diagnostic errors going forward.

2.
Cureus ; 14(6): e25768, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35706441

RESUMEN

Background Fluctuating asymmetry (FA) is widely defined as the deviation from perfect bilateral symmetry and is considered an epigenetic measure of environmental stress. Rinaldi and Fontani hypothesized that the FA morpho-functional changes originate from an adaptive motor behavior determined by functional alterations in the cerebellum and neural circuits, not caused by a lesion, but induced by environmental stress. They called this phenomenon functional dysmetria (FD). On this premise, they developed the radio electric asymmetric conveyer (REAC) technology, a neuromodulation technology aimed at optimizing the best neuro-psycho-motor strategies in relation to environmental interaction. Aims Previous studies showed that specific REAC neuro postural optimization (NPO) treatment can induce stable FD recovery. This study aimed to verify the duration of the NPO effect in inducing the stable FD recovery over time. Materials and methods Data were retrospectively collected from a population of 29,794 subjects who underwent a specific semiological FD assessment and received the NPO treatment, regardless of the pathology referred. Results The analysis of the data collected by the various participants in the study led us to ascertain the disappearance of FD in 100% of the cases treated, with a stability of the result detected up to 18 years after the single administration of the REAC NPO treatment. Conclusions The REAC NPO neurobiological modulation treatment consisting of a single administration surprisingly maintains a very long efficacy in the correction of FD. This effect can be explained as the long-lasting capacity of the NPO treatment to induce greater functional efficiency of the brain dynamics as proven in previous studies.

3.
Pediatrics ; 149(5)2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35490288

RESUMEN

BACKGROUND: Food insecurity (FI) increases children's risk for illness and developmental and behavioral problems, which are ongoing concerns for congenital heart disease (CHD) patients. In 2020, 14.8% of households with children suffered from FI. The Hunger Vital Signs (HVS) asks 2 questions to assess FI. The global aim of the project is to implement HVS and connect FI families to resources. METHODS: Stakeholders identified 6 critical drivers in implementing FI screening at an outpatient cardiology clinic and conducted plan-do-study-act (PDSA) cycles to implement HVS. Over the 13-month study period, time series analyses were performed to assess our process measure (FI screening) and outcome measure (connection of FI families to resources). Demographics and severity of CHD were analyzed for FI families. RESULTS: Screening rates increased from 0% to >85%, screening 5064 families. Process evaluations revealed roadblocks including screening discomfort. FI families were more likely to identify as Black or multiple or other ethnicity. Severe CHD patients were at higher risk for FI (n = 106, odds ratio [OR] 1.67 [1.21-2.29], P = .002). Face-to-face meetings with social work and community partnerships reduced loss to follow-up and our ability to offer all FI families individualized FI resources. CONCLUSION: HVS screening can be implemented in a cardiology clinic to improve identification of FI families. A written tool can combat screening discomfort and improve identification of FI families. Children with severe CHD may be at increased risk for FI. A multidisciplinary team and community partnerships can improve individualized resource distribution.


Asunto(s)
Cardiología , Abastecimiento de Alimentos , Instituciones de Atención Ambulatoria , Niño , Inseguridad Alimentaria , Humanos , Tamizaje Masivo
4.
Hum Mutat ; 30(11): 1567-73, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19760749

RESUMEN

The primary pathology in many cases of myelodysplasia (MDS) and acute myeloid leukemia (AML) remains unknown. In some cases, two or more affected members have been identified in the same family. To date, mutations in two genes have been directly implicated: the hematopoietic transcription factors RUNX1 (runt-related transcription factor 1) and CEBPA (CCATT-box enhancer binding protein alpha). However, there are also other familial cases of MDS/AML where the genetic basis remains unknown. Both MDS, and to a lesser extent AML, have been observed in cases of the bone marrow failure syndrome dyskeratosis congenita, in which telomerase mutations have been identified. Recently, an increased incidence of telomerase reverse transcriptase mutations has been reported in a series of de novo AML. We have now identified novel mutations in the telomerase RNA (TERC) or telomerase reverse transcriptase component (TERT) within 4 of 20 families presenting with familial MDS/AML. Functional analysis has demonstrated that all mutations adversely impact on telomerase activity in vitro, and affected individuals have short telomeres. These families, in conjunction with a review of previously published cases, help to further define the pathological role of telomerase mutations in MDS/AML and have implications for the biology, treatment and screening regimen of de novo cases.


Asunto(s)
Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicos/genética , ARN/genética , Telomerasa/genética , Secuencia de Aminoácidos , Femenino , Humanos , Masculino , Datos de Secuencia Molecular , Linaje
5.
Proc Natl Acad Sci U S A ; 105(23): 8073-8, 2008 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-18523010

RESUMEN

Dyskeratosis congenita is a premature aging syndrome characterized by muco-cutaneous features and a range of other abnormalities, including early greying, dental loss, osteoporosis, and malignancy. Dyskeratosis congenita cells age prematurely and have very short telomeres. Patients have mutations in genes that encode components of the telomerase complex (dyskerin, TERC, TERT, and NOP10), important in the maintenance of telomeres. Many dyskeratosis congenita patients remain uncharacterized. Here, we describe the analysis of two other proteins, NHP2 and GAR1, that together with dyskerin and NOP10 are key components of telomerase and small nucleolar ribonucleoprotein (snoRNP) complexes. We have identified previously uncharacterized NHP2 mutations that can cause autosomal recessive dyskeratosis congenita but have not found any GAR1 mutations. Patients with NHP2 mutations, in common with patients bearing dyskerin and NOP10 mutations had short telomeres and low TERC levels. SiRNA-mediated knockdown of NHP2 in human cells led to low TERC levels, but this reduction was not observed after GAR1 knockdown. These findings suggest that, in human cells, GAR1 has a different impact on the accumulation of TERC compared with dyskerin, NOP10, and NHP2. Most of the mutations so far identified in patients with classical dyskeratosis congenita impact either directly or indirectly on the stability of RNAs. In keeping with this effect, patients with dyskerin, NOP10, and now NHP2 mutations have all been shown to have low levels of telomerase RNA in their peripheral blood, providing direct evidence of their role in telomere maintenance in humans.


Asunto(s)
Envejecimiento Prematuro/genética , Disqueratosis Congénita/genética , Mutación/genética , Proteínas Nucleares/genética , Proteínas de Unión al ARN/genética , Ribonucleoproteínas Nucleares Pequeñas/genética , Telomerasa/genética , Sustitución de Aminoácidos , Secuencia de Bases , Análisis Mutacional de ADN , Femenino , Genes Recesivos , Células HeLa , Humanos , Masculino , Datos de Secuencia Molecular , Proteínas Mutantes/metabolismo , Proteínas Nucleares/química , Linaje , Estructura Secundaria de Proteína , Proteínas de Unión al ARN/química , Ribonucleoproteínas Nucleares Pequeñas/química , Ribonucleoproteínas Nucleolares Pequeñas/metabolismo , Síndrome , Telómero/metabolismo
6.
Nat Genet ; 40(5): 516-22, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18443588

RESUMEN

The rat is an important system for modeling human disease. Four years ago, the rich 150-year history of rat research was transformed by the sequencing of the rat genome, ushering in an era of exceptional opportunity for identifying genes and pathways underlying disease phenotypes. Genome-wide association studies in human populations have recently provided a direct approach for finding robust genetic associations in common diseases, but identifying the precise genes and their mechanisms of action remains problematic. In the context of significant progress in rat genomic resources over the past decade, we outline achievements in rat gene discovery to date, show how these findings have been translated to human disease, and document an increasing pace of discovery of new disease genes, pathways and mechanisms. Finally, we present a set of principles that justify continuing and strengthening genetic studies in the rat model, and further development of genomic infrastructure for rat research.


Asunto(s)
Modelos Animales de Enfermedad , Enfermedades Genéticas Congénitas/genética , Genoma , Genómica/tendencias , Ratas/genética , Animales , Animales Modificados Genéticamente , Mapeo Cromosómico , Marcación de Gen , Humanos
7.
Blood ; 110(13): 4198-205, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17785587

RESUMEN

Dyskeratosis congenita (DC) is a multisystem bone marrow failure syndrome characterized by a triad of mucocutaneous abnormalities and an increased predisposition to malignancy. X-linked DC is due to mutations in DKC1, while heterozygous mutations in TERC (telomerase RNA component) and TERT (telomerase reverse transcriptase) have been found in autosomal dominant DC. Many patients with DC remain uncharacterized, particularly families displaying autosomal recessive (AR) inheritance. We have now identified novel homozygous TERT mutations in 2 unrelated consanguineous families, where the index cases presented with classical DC or the more severe variant, Hoyeraal-Hreidarsson (HH) syndrome. These TERT mutations resulted in reduced telomerase activity and extremely short telomeres. As these mutations are homozygous, these patients are predicted to have significantly reduced telomerase activity in vivo. Interestingly, in contrast to patients with heterozygous TERT mutations or hemizygous DKC1 mutations, these 2 homozygous TERT patients were observed to have higher-than-expected TERC levels compared with controls. Collectively, the findings from this study demonstrate that homozygous TERT mutations, resulting in a pure but severe telomerase deficiency, produce a phenotype of classical AR-DC and its severe variant, the HH syndrome.


Asunto(s)
Disqueratosis Congénita/genética , Mutación , Telomerasa/genética , Disqueratosis Congénita/enzimología , Salud de la Familia , Homocigoto , Humanos , Fenotipo , Síndrome , Telomerasa/deficiencia
8.
Haematologica ; 92(8): 1013-20, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17640862

RESUMEN

BACKGROUND AND OBJECTIVES: Functional characterization of heterozygous TERC (telomerase RNA component) and TERT (telomerase reverse transcriptase) mutations found in autosomal dominant dyskeratosis congenita (DC) and aplastic anemia (AA) shows that telomerase function is defective and that this is associated with short telomeres. This leads to reduced cell longevity with maximal impact on tissues with high proliferate potential. The aim of this study was to establish the role of TERC in the pathophysiology of uncharacterized patients with AA with some features of DC. DESIGN AND METHODS: The TERC gene was screened for mutations by denaturing high performance liquid chromatography. To determine the functional significance of TERC mutations telomerase activity was assessed in an in vitro (TRAP) assay and telomere length of patients' samples was determined using Southern blot analysis. RESULTS This study led to the identification of four novel TERC mutations (G178A, C180T, D52-86 and G2C) and a recurrent TERC mutation (D110-113GACT). INTERPRETATION AND CONCLUSIONS: Two of the de novo TERC mutations (G178A and C180T) found uniquely produce a clinical phenotype in the first generation, differing from previously published cases in which individuals in the first generation are usually asymptomatic. Curiously these mutations are located near the triple-helix domain of TERC. We also observed that the recurrent D110-113GACT can present with AA, myelodysplasia or leukemia. The D52-86 is associated with varied phenotypes including pulmonary disease (pulmonary fibrosis) as the first presentation. In summary, this study reports the functional characterization of several novel TERC mutations associated with varied hematologic and extra-hematologic presentations.


Asunto(s)
Anemia Aplásica/genética , Disqueratosis Congénita/genética , Mutación , ARN/genética , Telomerasa/genética , Telómero/ultraestructura , Adolescente , Adulto , Secuencia de Bases , Preescolar , Femenino , Heterogeneidad Genética , Humanos , Discapacidad Intelectual/genética , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Nevo Pigmentado/genética , Conformación de Ácido Nucleico , Linaje , Fenotipo , Trastornos de la Pigmentación/genética , Fibrosis Pulmonar/genética , Telomerasa/deficiencia
9.
Hum Mol Genet ; 16(13): 1619-29, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17507419

RESUMEN

Dyskeratosis congenita (DC) is characterized by multiple features including mucocutaneous abnormalities, bone marrow failure and an increased predisposition to cancer. It exhibits marked clinical and genetic heterogeneity. DKC1 encoding dyskerin, a component of H/ACA small nucleolar ribonucleoprotein (snoRNP) particles is mutated in X-linked recessive DC. Telomerase RNA component (TERC), the RNA component and TERT the enzymatic component of telomerase, are mutated in autosomal dominant DC, suggesting that DC is primarily a disease of defective telomere maintenance. The gene(s) involved in autosomal recessive DC remains elusive. This paper describes studies aimed at defining the genetic basis of AR-DC. Homozygosity mapping in 16 consanguineous families with 25 affected individuals demonstrates that there is no single genetic locus for AR-DC. However, we show that NOP10, a component of H/ACA snoRNP complexes including telomerase is mutated in a large consanguineous family with classical DC. Affected homozygous individuals have significant telomere shortening and reduced TERC levels. While a reduction of TERC levels is not a universal feature of DC, it can be brought about through a reduction of NOP10 transcripts, as demonstrated by siRNA interference studies. A similar reduction in TERC levels is also seen when the mutant NOP10 is expressed in HeLa cells. These findings identify the genetic basis of one subtype of AR-DC being due to the first documented mutations in NOP10. This further strengthens the model that defective telomere maintenance is the primary pathology in DC and substantiates the evidence in humans for the involvement of NOP10 in the telomerase complex and telomere maintenance.


Asunto(s)
Disqueratosis Congénita/genética , Mutación , Ribonucleoproteínas Nucleolares Pequeñas/genética , Telomerasa/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Consanguinidad , Ligamiento Genético , Marcadores Genéticos , Células HeLa , Homocigoto , Humanos , Datos de Secuencia Molecular , Linaje , ARN Interferente Pequeño/metabolismo , Telómero/ultraestructura
10.
Blood ; 107(7): 2680-5, 2006 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-16332973

RESUMEN

The two genes mutated in the bone marrow failure syndrome dyskeratosis congenita (DC) both encode components of the telomerase complex responsible for maintaining the ends of chromosomes in stem cells and in the germ line. In reviewing the mutation profile that is found in DC, we describe 9 novel mutations in the DKC1 gene and 3 novel TERC mutations responsible for the X-linked and autosomal dominant forms of the disease, respectively, but find that two thirds of the families do not have mutations in either of these genes. In a significant subset of these uncharacterized families, the index case presents with severe disease previously defined as the Hoyeraal Hreidarsson (HH) syndrome. The diverse clinical phenotype seen in patients with X-linked DC is not explained by the different amino acid substitutions: Presentation of the recurrent A353V substitution ranges from classic DC to the severe HH variant. However, we do see that patients with HH have significantly shorter telomeres than those with a relatively mild presentation. In the new families described with TERC mutations, there is further evidence of disease anticipation associated with shorter telomeres in the younger generations. This study highlights the considerable genetic and phenotypic diversity of DC.


Asunto(s)
Disqueratosis Congénita/genética , Variación Genética , Mutación , ARN/genética , Telomerasa/genética , Telómero/genética , Cromosomas Humanos X , Femenino , Genes Dominantes , Humanos , Masculino , Modelos Moleculares , ARN/química , Hermanos , Telomerasa/química , Telómero/ultraestructura
11.
Int J Hematol ; 82(3): 184-9, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16207588

RESUMEN

Dyskeratosis congenita (DC) is a rare multisystem bone marrow failure syndrome that displays marked clinical and genetic heterogeneity. X-linked recessive, autosomal dominant and autosomal recessive forms of the disease are recognized. The gene that is mutated in the X-linked form of the disease is DKC1. The DKC1-encoded protein, dyskerin, is a component of small nucleolar ribonucleoprotein particles, which are important in ribosomal RNA processing, and of the telomerase complex. The autosomal dominant form of DC is due to mutations in the gene for the RNA component of telomerase (TERC). Because both dyskerin and TERC are components of the telomerase complex and all patients with DC have short telomeres, the principal pathology of DC appears to relate to telomerase dysfunction, although defects in ribosomal processing via dyskerin's involvement in pseudouridylation cannot be completely ruled out. The gene or genes involved in autosomal recessive DC remain elusive, although genes whose products are required for telomere maintenance remain strong candidates. The study of DC highlights the importance of telomerase in humans and how its deficiency results in multiple abnormalities, including premature aging, bone marrow failure, and cancer.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Disqueratosis Congénita/metabolismo , Proteínas Nucleares/metabolismo , Telómero/metabolismo , Proteínas de Ciclo Celular/genética , Cromosomas Humanos X/genética , Cromosomas Humanos X/metabolismo , Disqueratosis Congénita/genética , Disqueratosis Congénita/patología , Humanos , Proteínas Nucleares/genética , Procesamiento Postranscripcional del ARN/genética , ARN Ribosómico/genética , ARN Ribosómico/metabolismo , Telomerasa/genética , Telomerasa/metabolismo , Telómero/genética
12.
Blood Cells Mol Dis ; 34(3): 257-63, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15885610

RESUMEN

Human telomerase has two core components, the RNA molecule (TERC) that provides the template for telomere repeat elongation and a reverse transcriptase (TERT) that is responsible for the addition of telomere repeats at the ends of each chromosome. Mutations in TERC have been found in the autosomal-dominant form of the inherited bone marrow failure syndrome dyskeratosis congenita and in a subset of patients with aplastic anemia and myelodysplasia. These patients have short telomeres compared to age-matched controls. These observations suggest that uncharacterised cases of dyskeratosis congenita/aplastic anemia may have mutations in TERT or other molecules that associate with TERC in the telomerase complex. We have therefore screened the TERT gene for mutation by denaturing HPLC in 80 patients with inherited and acquired bone marrow failure (24 with dyskeratosis congenita, 36 with constitutional aplastic anemia, 13 with idiopathic aplastic anemia and 7 with other forms of bone marrow failure). 15 different TERT mutations have been identified. Of these, 5 are in flanking intron sequences, 6 are synonymous and 4 are non-synonymous (missense) substitutions in the coding sequence. These are the first natural mutations of TERT to be described and we highlight their possible pathogenic role in the development of bone marrow failure.


Asunto(s)
Enfermedades de la Médula Ósea/enzimología , Mutación , Adulto , Anemia Aplásica/enzimología , Anemia Aplásica/etiología , Anemia Aplásica/genética , Enfermedades de la Médula Ósea/etiología , Enfermedades de la Médula Ósea/genética , Niño , Preescolar , Análisis Mutacional de ADN , Proteínas de Unión al ADN/genética , Disqueratosis Congénita/enzimología , Disqueratosis Congénita/etiología , Disqueratosis Congénita/genética , Exones , Femenino , Humanos , Intrones , Masculino , Linaje , ADN Polimerasa Dirigida por ARN/genética , Telomerasa/genética
13.
Curr Opin Genet Dev ; 15(3): 249-57, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15917199

RESUMEN

Dyskeratosis congenita (DC) is a rare bone marrow failure syndrome that displays marked clinical and genetic heterogeneity. The identification of dyskeratosis congenita gene 1 (DKC1) mutations in X-linked recessive patients initially suggested that DC is a defective pseudouridylation disorder. The subsequent identification of mutations in the telomerase RNA component (TERC) of autosomal dominant DC patients together with the discovery that both TERC and the DKC1-encoded protein, dyskerin, are closely associated in the telomerase complex have suggested that the pathophysiology of DC predominantly relates to defective telomere maintenance. Recent discoveries have shown that autosomal dominant DC exhibits disease anticipation and that this is associated with progressive telomere shortening owing to the haplo-insufficiency of TERC.


Asunto(s)
Anticipación Genética/genética , Disqueratosis Congénita/enzimología , Disqueratosis Congénita/genética , Telomerasa/metabolismo , Telómero/genética , Telómero/metabolismo , Animales , Humanos , Mutación/genética
14.
Expert Rev Mol Med ; 6(26): 1-23, 2004 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-15613268

RESUMEN

Dyskeratosis congenita (DC) is a severe, inherited, bone marrow failure syndrome, with associated cutaneous and noncutaneous abnormalities. DC patients also show signs of premature ageing and have an increased occurrence of cancer. DC can originate through: (1) mutations in DKC1, which result in X-linked recessive DC; (2) mutations in the RNA component of telomerase (TERC), which result in autosomal dominant DC (AD-DC); and (3) mutations in other, currently uncharacterized, genes, which result in autosomal recessive DC (AR-DC). As DKC1 encodes dyskerin, a protein component of small nucleolar ribonucleoprotein (snoRNP) particles, which are important in ribosomal RNA processing, DC was initially described as a disorder of defective ribosomal biogenesis. Subsequently, dyskerin and TERC were shown to closely associate with each other in the telomerase complex, and DC has since come to be regarded as a telomerase deficiency disorder characterised by shorter telomeres. These findings demonstrate the importance of telomerase in humans and highlight how its deficiency (through DKC1 and TERC mutations) results in multiple abnormalities including premature ageing, bone marrow failure and cancer. Identification of the gene(s) involved in AR-DC will help to define the pathophysiology of DC further, as well as expand our insights into telomere function, ageing and cancer.


Asunto(s)
Envejecimiento Prematuro/genética , Envejecimiento/genética , Disqueratosis Congénita/genética , Neoplasias/genética , ARN/fisiología , Telomerasa/fisiología , Médula Ósea/fisiopatología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiología , Transformación Celular Neoplásica/genética , Disqueratosis Congénita/fisiopatología , Femenino , Heterogeneidad Genética , Humanos , Masculino , Mutación , Neoplasias/enzimología , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Conformación de Ácido Nucleico , ARN/química , ARN/genética , Procesamiento Postranscripcional del ARN , Ribonucleoproteínas Nucleolares Pequeñas/metabolismo , Telomerasa/química , Telomerasa/genética , Telómero/metabolismo , Telómero/ultraestructura
15.
Blood ; 104(13): 3936-42, 2004 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-15319288

RESUMEN

Mutations in TERC, encoding the RNA component of telomerase, have been found in autosomal dominant dyskeratosis congenita (DC) and aplastic anemia (AA). Several polymorphisms also exist in the TERC gene, making functional testing of potential pathogenic mutations essential. Here, we have tested normal and mutant TERC molecules in 2 telomerase reconstitution assays, 1 in vitro and 1 in transfected telomerase-negative cells. We find that 2 polymorphic mutations G58A and G228A have no effect on telomerase activity in these assays, whereas 6 mutations found in DC and AA cause reduction or abolition of telomerase activity. Mutations in the pseudoknot region of the TERC molecule, C72G, 96-7DeltaCT, GC107-8AG and 110-3DeltaGACT reduce the catalytic activity of reconstituted telomerase, whereas mutations in the 3' portion of the molecule C408G and a deletion of the 3' 74 bases have normal activity in vitro but reduced intracellular activity. By analyzing second site mutations that recreate regions of secondary structure but retain the pathogenic mutations we show that mutations C72G, GC107-8AG, and C408G act by disrupting the secondary structure or folding of TERC. Finally, experiments reconstituting telomerase with both normal and mutant TERC molecules suggest the mutations act via haploinsufficiency rather than by a dominant-negative mechanism.


Asunto(s)
Anemia Aplásica/genética , Disqueratosis Congénita/genética , Mutación , ARN/genética , Telomerasa/genética , Adulto , Anemia Aplásica/enzimología , Secuencia de Bases , Línea Celular , Niño , Disqueratosis Congénita/enzimología , Femenino , Tamización de Portadores Genéticos , Humanos , Recién Nacido , Masculino , Conformación de Ácido Nucleico , ARN/metabolismo , Eliminación de Secuencia , Telomerasa/metabolismo , Transfección
16.
BMC Blood Disord ; 4(1): 3, 2004 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-15212690

RESUMEN

BACKGROUND: Mutations in the gene coding for the RNA component of telomerase, hTERC, have been found in autosomal dominant dyskeratosis congenita (DC) and aplastic anemia. Paroxysmal nocturnal hemoglobinuria (PNH) is a clonal blood disorder associated with aplastic anemia and characterized by the presence of one or more clones of blood cells lacking glycosylphosphatidylinositol (GPI) anchored proteins due to a somatic mutation in the PIGA gene. METHODS: We searched for mutations in DNA extracted from PNH patients by amplification of the hTERC gene and denaturing high performance liquid chromatography (dHPLC). After a mutation was found in a potential transcription factor binding site in one patient electrophoretic mobility shift assays were used to detect binding of transcription factors to that site. The effect of the mutation on the function of the promoter was tested by transient transfection constructs in which the promoter is used to drive a reporter gene. RESULTS: Here we report the finding of a novel promoter mutation (-99C->G) in the hTERC gene in a patient with PNH. The mutation disrupts an Sp1 binding site and destroys its ability to bind Sp1. Transient transfection assays show that mutations in this hTERC site including C-99G cause either up- or down-regulation of promoter activity and suggest that the site regulates core promoter activity in a context dependent manner in cancer cells. CONCLUSIONS: These data are the first report of an hTERC promoter mutation from a patient sample which can modulate core promoter activity in vitro, raising the possibility that the mutation may affect the transcription of the gene in hematopoietic stem cells in vivo, and that dysregulation of telomerase may play a role in the development of bone marrow failure and the evolution of PNH clones.

17.
Nat Genet ; 36(5): 447-9, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15098033

RESUMEN

Telomerase is a ribonucleoprotein complex that is required to synthesize DNA repeats at the ends of each chromosome. The RNA component of this reverse transcriptase is mutated in the bone marrow failure syndrome autosomal dominant dyskeratosis congenita. Here we show that disease anticipation is observed in families with this disease and that this is associated with progressive telomere shortening.


Asunto(s)
Disqueratosis Congénita/genética , Mutación/genética , ARN/genética , Telomerasa/genética , Telómero/genética , Adolescente , Adulto , Niño , Preescolar , Disqueratosis Congénita/diagnóstico , Familia , Femenino , Genes Dominantes , Humanos , Masculino , Persona de Mediana Edad , Linaje , Eliminación de Secuencia
18.
Br J Haematol ; 119(3): 765-8, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12437656

RESUMEN

X-linked Hoyeraal-Hreidarsson syndrome (XL-HHS) is the severe infantile variant of X-linked dyskeratosis congenita (XL-DC) and both are due to mutations in the DKC1 gene within Xq28. We report a novel missense mutation in DKC1 exon 3 (T113-->C, Ile38Thr) in a Sardinian infant with XL-HHS in whom the disease was characterized by 'T+B-NK-' severe combined immunodeficiency and bone marrow failure. He underwent sibling bone marrow transplantation using a conditioning regimen (fludarabine, rabbit antithymocyte globulin, low-dose melphalan) selected according to the HHS/DC phenotype. This was associated with low toxicity, prompt engraftment with adequate immune reconstitution and full donor haemopoiesis.


Asunto(s)
Enfermedades de la Médula Ósea/genética , Trasplante de Médula Ósea/métodos , Proteínas de Ciclo Celular/genética , Disqueratosis Congénita/genética , Mutación Missense/genética , Proteínas Nucleares/genética , Inmunodeficiencia Combinada Grave/genética , Enfermedades de la Médula Ósea/terapia , Supervivencia de Injerto , Humanos , Lactante , Masculino , Inmunodeficiencia Combinada Grave/terapia , Síndrome , Acondicionamiento Pretrasplante/métodos
19.
Lancet ; 359(9324): 2168-70, 2002 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-12090986

RESUMEN

The main cause of aplastic anaemia remains elusive. Germline mutations in the gene encoding the RNA component of telomerase (hTR) have been seen in the autosomal dominant form of dyskeratosis congenita--an inherited syndrome characterised by aplastic anaemia. By screening the hTR gene, we identified mutations in two of 17 patients with idiopathic aplastic anaemia, three of 27 patients with constitutional aplastic anaemia, but in none of 214 normal controls (p<0.0001). Furthermore, patients with hTR mutations had significantly shorter telomeres than age-matched controls (p=0.027). These data indicate that, in a subset of patients with aplastic anaemia, the disorder might be associated with a genetic lesion in the telomere maintenance pathway.


Asunto(s)
Anemia Aplásica/genética , Reacción en Cadena de la Polimerasa/métodos , ARN/genética , Telomerasa/genética , Adolescente , Adulto , Anemia Aplásica/clasificación , Estudios de Casos y Controles , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Mutación/genética , ARN/aislamiento & purificación , Telomerasa/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...